Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Transplant ; 24(3): 350-361, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37931753

RESUMEN

The XVIth Banff Meeting for Allograft Pathology was held in Banff, Alberta, Canada, from September 19 to 23, 2022, as a joint meeting with the Canadian Society of Transplantation. In addition to a key focus on the impact of microvascular inflammation and biopsy-based transcript analysis on the Banff Classification, further sessions were devoted to other aspects of kidney transplant pathology, in particular T cell-mediated rejection, activity and chronicity indices, digital pathology, xenotransplantation, clinical trials, and surrogate endpoints. Although the output of these sessions has not led to any changes in the classification, the key role of Banff Working Groups in phrasing unanswered questions, and coordinating and disseminating results of investigations addressing these unanswered questions was emphasized. This paper summarizes the key Banff Meeting 2022 sessions not covered in the Banff Kidney Meeting 2022 Report paper and also provides an update on other Banff Working Group activities relevant to kidney allografts.


Asunto(s)
Trasplante de Riñón , Canadá , Rechazo de Injerto/etiología , Rechazo de Injerto/patología , Riñón/patología , Aloinjertos
4.
Transpl Int ; 36: 11589, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37680647

RESUMEN

The Thrombotic Microangiopathy Banff Working Group (TMA-BWG) was formed in 2015 to survey current practices and develop minimum diagnostic criteria (MDC) for renal transplant TMA (Tx-TMA). To generate consensus among pathologists and nephrologists, the TMA BWG designed a 3-Phase study. Phase I of the study is presented here. Using the Delphi methodology, 23 panelists with >3 years of diagnostic experience with Tx-TMA pathology listed their MDC suggesting light, immunofluorescence, and electron microscopy lesions, clinical and laboratory information, and differential diagnoses. Nine rounds (R) of consensus resulted in MDC validated during two Rs using online evaluation of whole slide digital images of 37 biopsies (28 TMA, 9 non-TMA). Starting with 338 criteria the process resulted in 24 criteria and 8 differential diagnoses including 18 pathologic, 2 clinical, and 4 laboratory criteria. Results show that 3/4 of the panelists agreed on the diagnosis of 3/4 of cases. The process also allowed definition refinement for 4 light and 4 electron microscopy lesions. For the first time in Banff classification, the Delphi methodology was used to generate consensus. The study shows that Delphi is a democratic and cost-effective method allowing rapid consensus generation among numerous physicians dealing with large number of criteria in transplantation.


Asunto(s)
Trasplante de Riñón , Microangiopatías Trombóticas , Humanos , Consenso , Análisis Costo-Beneficio , Biopsia
5.
Transpl Int ; 36: 11590, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37680648

RESUMEN

The Banff community summoned the TMA Banff Working Group to develop minimum diagnostic criteria (MDC) and recommendations for renal transplant TMA (Tx-TMA) diagnosis, which currently lacks standardized criteria. Using the Delphi method for consensus generation, 23 nephropathologists (panelists) with >3 years of diagnostic experience with Tx-TMA were asked to list light, immunofluorescence, and electron microscopic, clinical and laboratory criteria and differential diagnoses for Tx-TMA. Delphi was modified to include 2 validations rounds with histological evaluation of whole slide images of 37 transplant biopsies (28 TMA and 9 non-TMA). Starting with 338 criteria in R1, MDC were narrowed down to 24 in R8 generating 18 pathological, 2 clinical, 4 laboratory criteria, and 8 differential diagnoses. The panelists reached a good level of agreement (70%) on 76% of the validated cases. For the first time in Banff classification, Delphi was used to reach consensus on MDC for Tx-TMA. Phase I of the study (pathology phase) will be used as a model for Phase II (nephrology phase) for consensus regarding clinical and laboratory criteria. Eventually in Phase III (consensus of the consensus groups) and the final MDC for Tx-TMA will be reported to the transplantation community.


Asunto(s)
Trasplante de Riñón , Microangiopatías Trombóticas , Humanos , Trasplante de Riñón/efectos adversos , Consenso , Riñón , Microangiopatías Trombóticas/diagnóstico , Microangiopatías Trombóticas/etiología , Aminas , Anticoagulantes , Aloinjertos
6.
Nephrol Dial Transplant ; 37(12): 2351-2362, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-35772019

RESUMEN

Kidney dysplasia is one of the most frequent causes of chronic kidney failure in children. While dysplasia is a histological diagnosis, the term 'kidney dysplasia' is frequently used in daily clinical life without histopathological confirmation. Clinical parameters of kidney dysplasia have not been clearly defined, leading to imprecise communication amongst healthcare professionals and patients. This lack of consensus hampers precise disease understanding and the development of specific therapies. Based on a structured literature search, we here suggest a common basis for clinical, imaging, genetic, pathological and basic science aspects of non-obstructive kidney dysplasia associated with functional kidney impairment. We propose to accept hallmark sonographic findings as surrogate parameters defining a clinical diagnosis of dysplastic kidneys. We suggest differentiated clinical follow-up plans for children with kidney dysplasia and summarize established monogenic causes for non-obstructive kidney dysplasia. Finally, we point out and discuss research gaps in the field.


Asunto(s)
Enfermedades Renales , Insuficiencia Renal , Anomalías Urogenitales , Niño , Humanos , Riñón/patología , Enfermedades Renales/patología , Insuficiencia Renal/patología
7.
J Am Soc Nephrol ; 31(12): 2773-2792, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32938648

RESUMEN

BACKGROUND: The roles of asymptomatic hyperuricemia or uric acid (UA) crystals in CKD progression are unknown. Hypotheses to explain links between UA deposition and progression of CKD include that (1) asymptomatic hyperuricemia does not promote CKD progression unless UA crystallizes in the kidney; (2) UA crystal granulomas may form due to pre-existing CKD; and (3) proinflammatory granuloma-related M1-like macrophages may drive UA crystal-induced CKD progression. METHODS: MALDI-FTICR mass spectrometry, immunohistochemistry, 3D confocal microscopy, and flow cytometry were used to characterize a novel mouse model of hyperuricemia and chronic UA crystal nephropathy with granulomatous nephritis. Interventional studies probed the role of crystal-induced inflammation and macrophages in the pathology of progressive CKD. RESULTS: Asymptomatic hyperuricemia alone did not cause CKD or drive the progression of aristolochic acid I-induced CKD. Only hyperuricemia with UA crystalluria due to urinary acidification caused tubular obstruction, inflammation, and interstitial fibrosis. UA crystal granulomas surrounded by proinflammatory M1-like macrophages developed late in this process of chronic UA crystal nephropathy and contributed to the progression of pre-existing CKD. Suppressing M1-like macrophages with adenosine attenuated granulomatous nephritis and the progressive decline in GFR. In contrast, inhibiting the JAK/STAT inflammatory pathway with tofacitinib was not renoprotective. CONCLUSIONS: Asymptomatic hyperuricemia does not affect CKD progression unless UA crystallizes in the kidney. UA crystal granulomas develop late in chronic UA crystal nephropathy and contribute to CKD progression because UA crystals trigger M1-like macrophage-related interstitial inflammation and fibrosis. Targeting proinflammatory macrophages, but not JAK/STAT signaling, can attenuate granulomatous interstitial nephritis.


Asunto(s)
Hiperuricemia/complicaciones , Hiperuricemia/patología , Nefritis Intersticial/etiología , Nefritis Intersticial/patología , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología , Animales , Enfermedades Asintomáticas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Granuloma/etiología , Granuloma/metabolismo , Granuloma/patología , Hiperuricemia/metabolismo , Ratones , Nefritis Intersticial/sangre , Insuficiencia Renal Crónica/sangre
8.
Am J Transplant ; 20(9): 2318-2331, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32463180

RESUMEN

The XV. Banff conference for allograft pathology was held in conjunction with the annual meeting of the American Society for Histocompatibility and Immunogenetics in Pittsburgh, PA (USA) and focused on refining recent updates to the classification, advances from the Banff working groups, and standardization of molecular diagnostics. This report on kidney transplant pathology details clarifications and refinements to the criteria for chronic active (CA) T cell-mediated rejection (TCMR), borderline, and antibody-mediated rejection (ABMR). The main focus of kidney sessions was on how to address biopsies meeting criteria for CA TCMR plus borderline or acute TCMR. Recent studies on the clinical impact of borderline infiltrates were also presented to clarify whether the threshold for interstitial inflammation in diagnosis of borderline should be i0 or i1. Sessions on ABMR focused on biopsies showing microvascular inflammation in the absence of C4d staining or detectable donor-specific antibodies; the potential value of molecular diagnostics in such cases and recommendations for use of the latter in the setting of solid organ transplantation are presented in the accompanying meeting report. Finally, several speakers discussed the capabilities of artificial intelligence and the potential for use of machine learning algorithms in diagnosis and personalized therapeutics in solid organ transplantation.


Asunto(s)
Rechazo de Injerto , Trasplante de Riñón , Inteligencia Artificial , Rechazo de Injerto/diagnóstico , Riñón , Trasplante de Riñón/efectos adversos , Linfocitos T
9.
Am J Pathol ; 189(2): 248-257, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30472210

RESUMEN

Mitotic catastrophe (MC) is a major cause of podocyte loss in vitro and in vivo. We evaluated urine samples (n = 184 urine samples from diabetic patients; n = 41 patients) from diabetic patients and determined the presence of podocytes in the urine and studied their characteristics, specifically asking whether apoptosis versus MC is present. We also evaluated diabetic glomeruli in renal biopsy specimens by electron microscopy (n = 54). A battery of stains including the antibody to podocalyxin (PCX) were used. PCX and podocytes (PCX+podo) showed nuclear morphologies such as a i) mononucleated normal shape (8.7%), ii) large and abnormal shape (3.8%), iii) multinucleated with or without micronucleoli (31.2%), iv) mitotic spindles (8.2%), v) single nucleus and denucleation combined (10.3%), and vi) denucleation only (37.0%). Large size/abnormal shape, multinucleation, mitotic spindles, and a combination of single nucleus and denucleation were considered features of MC (53.5%). Dual staining of PCX+podo was positive for Glepp 1 (50%), whereas none of PCX+podo were positive for nephrin, podocin, leukocyte, or parietal epithelial cell markers (cytokeratin 8), annexin V, cleaved caspase-3, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling. Ten percent of PCX+podo were positive for phosphorylated vimentin. Electron microscopy identified cellular and nuclear podocyte changes characteristic of MC. The majority of urine podocytes in diabetic patients showed MC, not apoptosis. This noninvasive approach may be clinically useful in determining progressive diabetic nephropathy or response to therapeutic intervention.


Asunto(s)
Apoptosis , Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Mitosis , Podocitos , Anciano , Biomarcadores/metabolismo , Biopsia , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/orina , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Femenino , Humanos , Glomérulos Renales/metabolismo , Glomérulos Renales/ultraestructura , Masculino , Persona de Mediana Edad , Podocitos/metabolismo , Podocitos/ultraestructura
10.
J Am Soc Nephrol ; 30(10): 1857-1869, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31296606

RESUMEN

BACKGROUND: Serum oxalate levels suddenly increase with certain dietary exposures or ethylene glycol poisoning and are a well known cause of AKI. Established contributors to oxalate crystal-induced renal necroinflammation include the NACHT, LRR and PYD domains-containing protein-3 (NLRP3) inflammasome and mixed lineage kinase domain-like (MLKL) protein-dependent tubule necroptosis. These studies examined the role of a novel form of necrosis triggered by altered mitochondrial function. METHODS: To better understand the molecular pathophysiology of oxalate-induced AIK, we conducted in vitro studies in mouse and human kidney cells and in vivo studies in mice, including wild-type mice and knockout mice deficient in peptidylprolyl isomerase F (Ppif) or deficient in both Ppif and Mlkl. RESULTS: Crystals of calcium oxalate, monosodium urate, or calcium pyrophosphate dihydrate, as well as silica microparticles, triggered cell necrosis involving PPIF-dependent mitochondrial permeability transition. This process involves crystal phagocytosis, lysosomal cathepsin leakage, and increased release of reactive oxygen species. Mice with acute oxalosis displayed calcium oxalate crystals inside distal tubular epithelial cells associated with mitochondrial changes characteristic of mitochondrial permeability transition. Mice lacking Ppif or Mlkl or given an inhibitor of mitochondrial permeability transition displayed attenuated oxalate-induced AKI. Dual genetic deletion of Ppif and Mlkl or pharmaceutical inhibition of necroptosis was partially redundant, implying interlinked roles of these two pathways of regulated necrosis in acute oxalosis. Similarly, inhibition of mitochondrial permeability transition suppressed crystal-induced cell death in primary human tubular epithelial cells. PPIF and phosphorylated MLKL localized to injured tubules in diagnostic human kidney biopsies of oxalosis-related AKI. CONCLUSIONS: Mitochondrial permeability transition-related regulated necrosis and necroptosis both contribute to oxalate-induced AKI, identifying PPIF as a potential molecular target for renoprotective intervention.


Asunto(s)
Lesión Renal Aguda/patología , Necrosis por Permeabilidad de la Transmembrana Mitocondrial , Necroptosis , Lesión Renal Aguda/inducido químicamente , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Oxalatos/administración & dosificación
11.
J Am Soc Nephrol ; 28(6): 1753-1768, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28073931

RESUMEN

Severe AKI is often associated with multiorgan dysfunction, but the mechanisms of this remote tissue injury are unknown. We hypothesized that renal necroinflammation releases cytotoxic molecules that may cause remote organ damage. In hypoxia-induced tubular epithelial cell necrosis in vitro, histone secretion from ischemic tubular cells primed neutrophils to form neutrophil extracellular traps. These traps induced tubular epithelial cell death and stimulated neutrophil extracellular trap formation in fresh neutrophils. In vivo, ischemia-reperfusion injury in the mouse kidney induced tubular necrosis, which preceded the expansion of localized and circulating neutrophil extracellular traps and the increased expression of inflammatory and injury-related genes. Pretreatment with inhibitors of neutrophil extracellular trap formation reduced kidney injury. Dual inhibition of neutrophil trap formation and tubular cell necrosis had an additive protective effect. Moreover, pretreatment with antihistone IgG suppressed ischemia-induced neutrophil extracellular trap formation and renal injury. Renal ischemic injury also increased the levels of circulating histones, and we detected neutrophil infiltration and TUNEL-positive cells in the lungs, liver, brain, and heart along with neutrophil extracellular trap accumulation in the lungs. Inhibition of neutrophil extracellular trap formation or of circulating histones reduced these effects as well. These data suggest that tubular necrosis and neutrophil extracellular trap formation accelerate kidney damage and remote organ dysfunction through cytokine and histone release and identify novel molecular targets to limit renal necroinflammation and multiorgan failure.


Asunto(s)
Lesión Renal Aguda/complicaciones , Trampas Extracelulares/fisiología , Isquemia/complicaciones , Necrosis de la Corteza Renal/etiología , Riñón/irrigación sanguínea , Neutrófilos , Animales , Células Cultivadas , Histonas/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Índice de Severidad de la Enfermedad
12.
BMC Nephrol ; 18(1): 57, 2017 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-28183278

RESUMEN

BACKGROUND: Castleman's disease (CD) is an uncommon, heterogeneous lympho-proliferative disorder leading to high circulating levels of interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF). Renal involvement has been only described in a limited number of small studies. Herein, we report a rare case of renal thrombotic microangiopathy (TMA) associated with CD and investigate the podocyte expression of VEGF in the renal biopsy prior to initiation of treatment. CASE PRESENTATION: An 18-year-old male presented with fever, diarrhea, diffuse lymphadenopathy, ascites and acute kidney injury. Laboratory tests for hemolytic uremic syndrome and thrombotic thrombocytopenic purpura were negative. The kidney biopsy showed TMA. An excisional lymph node biopsy was consistent with CD, plasma cell variant. Immunofluorescence staining showed suppressed podocyte VEGF expression. Chemotherapy that inhibits production of inflammatory mediators including IL-6 and VEGF led to complete recovery of renal function. CONCLUSIONS: Our case illustrates a rare renal histological feature of CD. IL-6 and VEGF are postulated to suppress glomerular VEGF expression, thereby causing renal TMA. Therapy directed against these inflammatory mediators may have important therapeutic implications.


Asunto(s)
Enfermedad de Castleman/diagnóstico , Enfermedad de Castleman/inmunología , Enfermedades Renales/diagnóstico , Enfermedades Renales/inmunología , Podocitos/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología , Adolescente , Biomarcadores/metabolismo , Enfermedad de Castleman/complicaciones , Diagnóstico Diferencial , Humanos , Enfermedades Renales/etiología , Masculino , Enfermedades Raras/diagnóstico por imagen , Microangiopatías Trombóticas
13.
J Am Soc Nephrol ; 27(6): 1635-49, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26567242

RESUMEN

Endothelial dysfunction is a central pathomechanism in diabetes-associated complications. We hypothesized a pathogenic role in this dysfunction of cathepsin S (Cat-S), a cysteine protease that degrades elastic fibers and activates the protease-activated receptor-2 (PAR2) on endothelial cells. We found that injection of mice with recombinant Cat-S induced albuminuria and glomerular endothelial cell injury in a PAR2-dependent manner. In vivo microscopy confirmed a role for intrinsic Cat-S/PAR2 in ischemia-induced microvascular permeability. In vitro transcriptome analysis and experiments using siRNA or specific Cat-S and PAR2 antagonists revealed that Cat-S specifically impaired the integrity and barrier function of glomerular endothelial cells selectively through PAR2. In human and mouse type 2 diabetic nephropathy, only CD68(+) intrarenal monocytes expressed Cat-S mRNA, whereas Cat-S protein was present along endothelial cells and inside proximal tubular epithelial cells also. In contrast, the cysteine protease inhibitor cystatin C was expressed only in tubules. Delayed treatment of type 2 diabetic db/db mice with Cat-S or PAR2 inhibitors attenuated albuminuria and glomerulosclerosis (indicators of diabetic nephropathy) and attenuated albumin leakage into the retina and other structural markers of diabetic retinopathy. These data identify Cat-S as a monocyte/macrophage-derived circulating PAR2 agonist and mediator of endothelial dysfunction-related microvascular diabetes complications. Thus, Cat-S or PAR2 inhibition might be a novel strategy to prevent microvascular disease in diabetes and other diseases.


Asunto(s)
Catepsinas/fisiología , Angiopatías Diabéticas/etiología , Células Endoteliales/enzimología , Receptor PAR-2/metabolismo , Animales , Catepsinas/antagonistas & inhibidores , Células Cultivadas , Glomérulos Renales/citología , Masculino , Ratones , Microvasos , Prolina/análogos & derivados , Prolina/farmacología , Urotelio/citología
14.
Lab Invest ; 96(4): 378-90, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26752746

RESUMEN

Acute kidney injury (AKI) involves nephron injury leading to irreversible nephron loss, ie, chronic kidney disease (CKD). Both AKI and CKD are associated with distinct histological patterns of tissue injury, but kidney atrophy in CKD involves tissue remodeling with interstitial inflammation and scarring. No doubt, nephron atrophy, inflammation, fibrosis, and renal dysfunction are associated with each other, but their hierarchical relationships remain speculative. To better understand the pathophysiology, we provide an overview of the fundamental danger response programs that assure host survival upon traumatic injury from as early as the first multicellular organisms, ie, bleeding control by coagulation, infection control by inflammation, epithelial barrier restoration by re-epithelialization, and tissue stabilization by mesenchymal repair. Although these processes assure survival in the majority of the populations, their dysregulation causes kidney disease in a minority. We discuss how, in genetically heterogeneous population, genetic variants shift balances and modulate danger responses toward kidney disease. We further discuss how classic kidney disease entities develop from an insufficient or overshooting activation of these danger response programs. Finally, we discuss molecular pathways linking, for example, inflammation and regeneration or inflammation and fibrosis. Understanding the causative and hierarchical relationships and the molecular links between the danger response programs should help to identify molecular targets to modulate kidney injury and to improve outcomes for kidney disease patients.


Asunto(s)
Coagulación Sanguínea/fisiología , Inflamación/fisiopatología , Riñón/fisiopatología , Regeneración/fisiología , Lesión Renal Aguda/fisiopatología , Epitelio/patología , Epitelio/fisiopatología , Fibrosis/fisiopatología , Humanos , Riñón/patología , Modelos Biológicos , Insuficiencia Renal Crónica/fisiopatología
15.
J Am Soc Nephrol ; 26(7): 1513-23, 2015 07.
Artículo en Inglés | MEDLINE | ID: mdl-25349197

RESUMEN

Murine double minute-2 (MDM2), an E3 ligase that regulates the cell cycle and inflammation, is highly expressed in podocytes. In podocyte injury, MDM2 drives podocyte loss by mitotic catastrophe, but the function of MDM2 in resting podocytes has not been explored. Here, we investigated the effects of podocyte MDM2 deletion in vitro and in vivo. In vitro, MDM2 knockdown by siRNA caused increased expression of p53 and podocyte death, which was completely rescued by coknockdown of p53. Apoptosis, pyroptosis, pyronecrosis, necroptosis, ferroptosis, and parthanatos were excluded as modes of occurrence for this p53-overactivation-related cell death (here referred to as podoptosis). Podoptosis was associated with cytoplasmic vacuolization, endoplasmic reticulum stress, and dysregulated autophagy (previously described as paraptosis). MDM2 knockdown caused podocyte loss and proteinuria in a zebrafish model, which was consistent with the phenotype of podocyte-specific MDM2-knockout mice that also showed the aforementioned ultrastructual podocyte abnormalities before and during progressive glomerulosclerosis. The phenotype of both animal models was entirely rescued by codeletion of p53. We conclude that MDM2 maintains homeostasis and long-term survival in podocytes by preventing podoptosis, a p53-regulated form of cell death with unspecific features previously classified as paraptosis.


Asunto(s)
Autofagia/genética , Muerte Celular/genética , Genes p53/fisiología , Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Activación Transcripcional/genética , Análisis de Varianza , Animales , Western Blotting , Supervivencia Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Genes p53/genética , Glomeruloesclerosis Focal y Segmentaria/patología , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Homeostasis/genética , Inmunohistoquímica , Pruebas de Función Renal , Ratones , Ratones Noqueados , Microscopía Confocal , Podocitos/citología , Podocitos/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Sensibilidad y Especificidad , Activación Transcripcional/fisiología , Pez Cebra
16.
J Am Soc Nephrol ; 26(10): 2399-413, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25644111

RESUMEN

Severe GN involves local neutrophil extracellular trap (NET) formation. We hypothesized a local cytotoxic effect of NET-related histone release in necrotizing GN. In vitro, histones from calf thymus or histones released by neutrophils undergoing NETosis killed glomerular endothelial cells, podocytes, and parietal epithelial cells in a dose-dependent manner. Histone-neutralizing agents such as antihistone IgG, activated protein C, or heparin prevented this effect. Histone toxicity on glomeruli ex vivo was Toll-like receptor 2/4 dependent, and lack of TLR2/4 attenuated histone-induced renal thrombotic microangiopathy and glomerular necrosis in mice. Anti-glomerular basement membrane GN involved NET formation and vascular necrosis, whereas blocking NET formation by peptidylarginine inhibition or preemptive anti-histone IgG injection significantly reduced all aspects of GN (i.e., vascular necrosis, podocyte loss, albuminuria, cytokine induction, recruitment or activation of glomerular leukocytes, and glomerular crescent formation). To evaluate histones as a therapeutic target, mice with established GN were treated with three different histone-neutralizing agents. Anti-histone IgG, recombinant activated protein C, and heparin were equally effective in abrogating severe GN, whereas combination therapy had no additive effects. Together, these results indicate that NET-related histone release during GN elicits cytotoxic and immunostimulatory effects. Furthermore, neutralizing extracellular histones is still therapeutic when initiated in established GN.


Asunto(s)
Vasos Sanguíneos/patología , Trampas Extracelulares/fisiología , Glomerulonefritis/complicaciones , Histonas/fisiología , Animales , Bovinos , Trampas Extracelulares/efectos de los fármacos , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/etiología , Histonas/efectos de los fármacos , Ratones , Necrosis/etiología , Índice de Severidad de la Enfermedad
17.
J Urol ; 193(5 Suppl): 1860-1869, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25813565

RESUMEN

PURPOSE: Congenital urinary tract obstruction is a leading cause of renal maldevelopment and pediatric kidney disease. Nonetheless, few groups have examined its molecular pathogenesis in humans. We evaluated the role of BMP-7, a protein required for renal injury repair and nephrogenesis, in disease progression in patients with obstructive uropathy. MATERIALS AND METHODS: Whole kidney and cell specific BMP-7 expression was examined in a murine model of unilateral ureteral obstruction and in patients with congenital ureteropelvic junction obstruction. Findings were correlated with molecular markers of renal injury and clinical parameters. RESULTS: Unilateral ureteral obstruction led to a dramatic decrease in BMP-7 expression in the proximal and distal tubules before the onset of significant loss of renal architecture and fibrosis, suggesting that this is a critical molecular event that drives early stage disease progression. Loss of BMP-7 expression then extended to the collecting ducts and glomeruli in end stage kidney disease. When translating these findings to patients with ureteropelvic junction obstruction, global loss of BMP-7 expression correlated with a decreased number of nephrons, loss of renal architecture, severe renal fibrosis and loss of kidney function. CONCLUSIONS: Given that BMP-7 has a critical role in renal injury repair and nephrogenesis, these findings show that cell specific changes in BMP-7 expression contribute to the onset of irreversible renal injury and impaired kidney development secondary to congenital urinary tract obstruction. Accordingly therapies that target these cell populations to restore BMP-7 activity may limit disease progression in patients with obstructive uropathy.


Asunto(s)
Proteína Morfogenética Ósea 7/metabolismo , Enfermedades Renales/metabolismo , Túbulos Renales Distales/metabolismo , Túbulos Renales Proximales/metabolismo , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Túbulos Renales Distales/citología , Túbulos Renales Proximales/citología , Ratones Endogámicos C57BL
18.
BMC Nephrol ; 16: 101, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26156092

RESUMEN

Recent advances show that human focal segmental glomerulosclerosis (FSGS) is a primary podocytopathy caused by podocyte-specific gene mutations including NPHS1, NPHS2, WT-1, LAMB2, CD2AP, TRPC6, ACTN4 and INF2. This review focuses on genes discovered in the investigation of complex FSGS pathomechanisms that may have implications for the current FSGS classification scheme. It also recounts recent recommendations for clinical management of FSGS based on translational studies and clinical trials. The advent of next-generation sequencing promises to provide nephrologists with rapid and novel approaches for the diagnosis and treatment of FSGS. A stratified and targeted approach based on the underlying molecular defects is evolving.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Inmunosupresores/uso terapéutico , Plasmaféresis , Actinina/genética , Adalimumab/uso terapéutico , Proteínas Adaptadoras Transductoras de Señales/genética , Corticoesteroides/uso terapéutico , Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Proteínas del Citoesqueleto/genética , Forminas , Glomeruloesclerosis Focal y Segmentaria/terapia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Factores Inmunológicos/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/genética , Laminina/genética , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Terapia Molecular Dirigida , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapéutico , Rituximab/uso terapéutico , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6 , Proteínas WT1/genética
19.
Am J Pathol ; 183(5): 1364-1374, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24007883

RESUMEN

Podocytes represent an essential component of the kidney's glomerular filtration barrier. They stay attached to the glomerular basement membrane via integrin interactions that support the capillary wall to withstand the pulsating filtration pressure. Podocyte structure is maintained by a dynamic actin cytoskeleton. Terminal differentiation is coupled with permanent exit from the cell cycle and arrest in a postmitotic state. Postmitotic podocytes do not have an infinite life span; in fact, physiologic loss in the urine is documented. Proteinuria and other injuries accelerate podocyte loss or induce death. Mature podocytes are unable to replicate and maintain their actin cytoskeleton simultaneously. By the end of mitosis, cytoskeletal actin forms part of the contractile ring, rendering a round shape to podocytes. Therefore, when podocyte mitosis is attempted, it may lead to aberrant mitosis (ie, mitotic catastrophe). Mitotic catastrophe implies that mitotic podocytes eventually detach or die; this is a previously unrecognized form of podocyte loss and a compensatory mechanism for podocyte hypertrophy that relies on post-G1-phase cell cycle arrest. In contrast, local podocyte progenitors (parietal epithelial cells) exhibit a simple actin cytoskeleton structure and can easily undergo mitosis, supporting podocyte regeneration. In this review we provide an appraisal of the in situ pathology of mitotic catastrophe compared with other proposed types of podocyte death and put experimental and renal biopsy data in a unified perspective.


Asunto(s)
Mitosis , Podocitos/patología , Animales , Muerte Celular , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Humanos , Hipertrofia , Enfermedades Renales/complicaciones , Enfermedades Renales/patología , Enfermedades Renales/virología , Podocitos/ultraestructura
20.
Am J Pathol ; 183(2): 431-40, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23747509

RESUMEN

Interferon (IFN)-α and IFN-ß are the central regulators of antiviral immunity but little is known about their roles in viral glomerulonephritis (eg, HIV nephropathy). We hypothesized that IFN-α and IFN-ß would trigger local inflammation and podocyte loss. We found that both IFNs consistently activated human and mouse podocytes and parietal epithelial cells to express numerous IFN-stimulated genes. However, only IFN-ß significantly induced podocyte death and increased the permeability of podocyte monolayers. In contrast, only IFN-α caused cell-cycle arrest and inhibited the migration of parietal epithelial cells. Both IFNs suppressed renal progenitor differentiation into mature podocytes. In Adriamycin nephropathy, injections with either IFN-α or IFN-ß aggravated proteinuria, macrophage influx, and glomerulosclerosis. A detailed analysis showed that only IFN-ß induced podocyte mitosis. This did not, however, lead to proliferation, but was associated with podocyte loss via podocyte detachment and/or mitotic podocyte death (mitotic catastrophe). We did not detect TUNEL-positive podocytes. Thus, IFN-α and IFN-ß have both common and differential effects on podocytes and parietal epithelial cells, which together promote glomerulosclerosis by enhancing podocyte loss while suppressing podocyte regeneration from local progenitors.


Asunto(s)
Antivirales/farmacología , Glomerulonefritis/tratamiento farmacológico , Interferón-alfa/farmacología , Interferón beta/farmacología , Animales , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Doxorrubicina/toxicidad , Células Epiteliales/efectos de los fármacos , Femenino , Glomerulonefritis/fisiopatología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/fisiopatología , Humanos , Glomérulos Renales/fisiología , Ratones , Ratones SCID , Podocitos/efectos de los fármacos , Regeneración/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA